You are on page 1of 9

This article was downloaded by: [178.131.172.

15]
On: 24 July 2015, At: 12:25
Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG

Cell Cycle
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/kccy20

Protective roles of SIRT1 in atherosclerosis


Sokrates Stein & Christian M. Matter
Published online: 15 Feb 2011.

To cite this article: Sokrates Stein & Christian M. Matter (2011) Protective roles of SIRT1 in atherosclerosis , Cell Cycle, 10:4,
640-647, DOI: 10.4161/cc.10.4.14863

To link to this article: http://dx.doi.org/10.4161/cc.10.4.14863

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
review

Cell Cycle 10:4, 640-647; February 15, 2011; © 2011 Landes Bioscience

Protective roles of SIRT1 in atherosclerosis


Sokrates Stein and Christian M. Matter
Cardiovascular Research; Institute of Physiology; Zurich Center for Integrative Human Physiology (ZIHP); University of Zurich; Zurich, Switzerland;
and Division of Cardiology; Cardiovascular Center; University Hospital Zurich; Zurich, Switzerland

Key words: sirtuin, SIRT1, atherosclerosis, thrombosis, inflammation, NFkB

Abbreviations: EC, endothelial cell; eNOS, endothelial nitric oxide synthase; HAEC, human aortic endothelial cell; ICAM-1,
intercellular adhesion molecule 1; LDL, low-density lipoprotein; Lox-1, lectin-like oxidized LDL receptor 1 (also called Olr1);
LXRα/β, liver x receptor; NAD, nicotinamide adenine dinucleotide; NAM, nicotinamide; NFκB, nuclear factor kappaB;
NO, nitric oxide; oxLDL, oxidized LDL; PGC-1α, PPARγ coactivator 1α; PPARγ, peroxisome proliferator-activated receptor γ;
ROS, reactive oxygen species; SR-A, scavenger receptor A; SR-B, scavenger receptor B; SR-PSOX/CXCL16, scavenger receptor for
phosphatidylserine and oxidized lipoprotein/CXC chemokine ligand 16; TF, tissue factor (also called coagulation factor III or F3);
TIMP3, tissue inhibitor of metalloproteinase 3; TNF-α, tumor necrosis factor alpha; VCAM-1, vascular cell adhesion molecule 1;
VSMC, vascular smooth muscle cell
Downloaded by [178.131.172.15] at 12:25 24 July 2015

SIRT1 is a NAD+-dependent class III histone deacetylase (HDAC) Sirtuins Deacetylate Nε-Lysines
that mediates the effects of caloric restriction on lifespan and
metabolic pathways in various organisms. It deacetylates both HDACs promote the deacetylation of acetyl-Nε-lysine residues
histone and non-histone proteins and targets proteins with and thereby affect functional properties of the target protein.1
diverse cellular and tissue functions. In the vasculature of rodent HDACs are discriminated into four families: classes I–IV. Class
models SIRT1 mediates vasodilatation via eNOS-derived nitric
©2011L
andesBiosc
ienc
e.
III HDACs are also called sirtuins. The mammalian sirtuin
oxide (NO) and scavenging reactive oxygen species (ROS). family consists of seven HDACs that share a conserved catalytic
Recent studies demonstrated further protective roles of SIRT1 core domain and are expressed ubiquitously.2 Sirtuins reverse
in vascular biology and atherosclerosis. In endothelial cells

Donotdi
str
ibut
e.
and macrophages SIRT1 has anti-inflammatory functions by
downregulating the expression of various pro-inflammatory
Nε-lysine acetylations of their target proteins by hydrolyzing one
NAD + and releasing nicotinamide (NAM), a unique byproduct
called O-acetyl-ADP-ribose (OAADPr) and the deacetylated
cytokines by interfering with the NFkB signaling pathway.
Deacetylation of RelA/p65-NFkB by SIRT1 in macrophages substrate.3-5 Sir2, the yeast homolog of SIRT1, was discovered
also suppresses the expression of Lox-1, a scavenger receptor as a factor necessary for the transcriptional silencing of the silent
for oxidized low-density lipoproteins (oxLDL), thereby mating-type loci and telomeric areas in yeast by inducing DNA
preventing macrophage foam cell formation. Moreover, SIRT1 heterochromatization.6-8 Homologs are also found in many
has been shown to regulate the activity of Liver X-receptor other species such as Escherichia coli, Salmonella typhimurium,
(LXR), thereby promoting ABCA1-driven reverse cholesterol Caenorhabditis elegans, Drosophila melanogaster, Mus musculus,
transport in plaque macrophages. Finally, SIRT1 suppresses the Macaca mulatta and Homo sapiens, highlighting the fact that the
expression of endothelial tissue factor (coagulation factor III) Sir2 family is conserved from bacteria to humans.9-12
and hence exerts anti-thrombotic properties. These findings
The finding that extra copies of Sir2 prolong the life span
indicate atheroprotective effects of SIRT1 in atherogenesis
and highlight the need for translational research from
of yeast, whereas Sir2 deficiency caused the opposite,13 drew the
bench-to-bedside. Indeed, SIRT1 activators are available for attention of many scientists to the Sir2/sirtuin field. It was shown
experimental research and undergo clinical testing. Taken that Sir2 is a NAD + -dependent histone deacetylase and that both
together, these studies suggest SIRT1 activation as a promising Sir2 and NAD + are required for calorie restriction-mediated life
therapeutic approach in atherosclerosis. Further studies are span extension in yeast.14-17 These findings placed Sir2 into a cen-
necessary to better understand the exact role of SIRT1 in the tral position linking the metabolic state of a cell with the tran-
protagonist cells orchestrating atherogenesis and to identify scriptional control of gene expression.
the specificity, target effects and putative off-target effects of Sir2 homologs and Sir2 activators also regulate the life span
these promising SIRT1 activators. in Caenorhabditis elegans and Drosophila melanogaster,18-20 and
it was recently demonstrated that moderate calorie restriction
lowers the incidence of aging-related deaths in rhesus mon-
keys.21 Nevertheless, it remains unclear whether sirtuins are
directly involved in calorie restriction-mediated lifespan exten-
Correspondence to: Sokrates Stein and Christian M. Matter; sion.22-25 On the other hand, the central role of SIRT1 as a
Email: sokrates@access.uzh.ch and christian.matter@access.uzh.ch metabolic regulator remains undisputed. Indeed, the function
Submitted: 01/11/11; Accepted: 01/19/11 of many important proteins involved in metabolism is regu-
DOI: 10.4161/cc.10.4.14863
lated by SIRT1, including peroxisome proliferator-activated

640 Cell Cycle Volume 10 Issue 4


review REVIEW

receptor γ (PPARγ), PPARγ coactivator 1α (PGC-1α), nuclear explain this discrepancy: (1) Chen et al. elegantly show the syner-
factor κB (NFκB), Liver X receptor α (LXRα or Nr1h3) gistic activation of eNOS by SIRT1 and AMPK. However, there
and endothelial nitric oxide synthase (eNOS or NOS3).26-31 is no genetic evidence derived from SIRT1 knockout cells or
Interestingly, many of these molecules have been found to play mice demonstrating that SIRT1 is essential to activate eNOS.37
a direct or indirect role in atherogenesis.32-35 The main findings Miyazaki used resveratrol to activate and nicotinamide to inhibit
regarding the direct function of SIRT1 in atherosclerosis will SIRT1 function. Of note, resveratrol is not a direct activator of
be discussed below. SIRT1 and nicotinamide affects the function of several sirtuins
and other proteins.47-49 Again, no confirmatory genetic studies
Laminar Shear Stress Induces SIRT1 Expression were performed.44 Mattagajasingh overexpressed a dominant-
negative SIRT1 mutant in rat aortic rings ex vivo. The study
Depending on the flow pattern in an artery, shear stress exerts is well conducted, and it would be interesting to extend it to
atheroprotective or atheroprone properties. While steady or pul- functional in vivo assays using pharmacological approaches.27
satile laminar flow with a clear direction confers anti-atherogenic Zhang et al. overexpressed the human SIRT1 gene in endothe-
effects by increasing the eNOS-derived NO bioavailability, a dis- lial cells of atherosclerotic mice, resulting in an increased eNOS
turbed turbulent flow is atheroprone by activating pro-inflam- expression and improved vascular function, but eNOS activity
matory and proliferative signaling cascades as well as increasing or phosphorylation was not assessed.45 Stein et al. used ApoE-/-
the level of ROS.36 Human umbilical vein endothelial cells SIRT1+/+ and haploinsufficient ApoE-/- SIRT1+/- mice to analyze
(HUVECs) exposed to laminar flow in vitro enhance the expres- the role of endogenous SIRT1 on vascular function. Although
sion of AMPK and SIRT1 and display increased NAD + levels and the intact allele cannot compensate the lack of SIRT1 transcript
Downloaded by [178.131.172.15] at 12:25 24 July 2015

SIRT1 activity. Consequently, AMPK-mediated eNOS phos- from the deficient allele in the haploinsufficient animals, the pos-
phorylation primes eNOS for deacetylation by SIRT1, thereby sibility that the single allele produces sufficient protein to regu-
activating eNOS and inducing NO generation.37 Consistently, late eNOS cannot be excluded.46 Therefore, further genetic and
expression of SIRT1 is higher in vessel areas under pulsatile flow pharmacological studies are necessary determine the impact of
(thoracic aorta) than in areas under disturbed flow (aortic arch) SIRT1 activity on eNOS and endothelium-dependent vascular

©2011L
andesBiosc
ienc
e.
in C57BL/6 mice.37 function.50

SIRT1 Improves Endothelial Function SIRT1 Inhibits Endothelial Cell Activation

Donotdi
str
ibut
e.
Loss of intact endothelial function over time may lead to chronic
endothelial dysfunction, which is an indicator of cardiovascu-
Cytokines (e.g., TNF-α and several interleukins) and constitu-
ents of modified lipoproteins (e.g., lysophosphatidyl choline)
lar disease and an early step in atherogenesis.38 Excessive pro- enhance oxidative stress in endothelial cells, which in turn acti-
duction of oxidant stress, which is often observed in patients vates the NFκB signaling pathway. Activated NFκB migrates
with hypertension, hypercholesterolemia or diabetes, impairs into the nucleus and induces the expression of numerous genes,
NO production and inactivates NO to form toxic peroxynitrite including many pro-inflammatory genes such as P-Selectin,
(ONOO-).39 Peroxynitrite may ‘uncouple’ eNOS, which means ICAM-1 and VCAM-1.51,52 These endothelial cells are consid-
that its oxygen reduction capacity is uncoupled from NO syn- ered ‘activated’. In contrast, under physiological conditions aortic
thesis, thereby yielding a dysfunctional superoxide-generating endothelial cells do not express these adhesion molecules, except
enzyme that enhances vascular oxidative stress.40 These and for low constitutive levels of ICAM-1.53 Activated endothelial
several other consequences of impaired NO biosynthesis dete- cells that present these molecules and secrete them into the blood
riorate endothelial function and induce sustained endothelial stream may recruit and activate blood monocytes, which then
inflammatory activation that contributes to the development of transmigrate into the arterial intima. This transmigration of
atherosclerosis.38,41-43 monocytes is stimulated by chemokines such as monocyte che-
Several reports proposed that SIRT1 improves endothelium- moattractant protein-1 (MCP‑1), MCP-4, regulated on activa-
dependent vascular function: as mentioned above, laminar shear tion normal T-cell expressed and secreted (RANTES) and IL-8,
stress enhances SIRT1 activity and activates eNOS.37 SIRT1 which bind to G protein-coupled receptors and are expressed by
activation by resveratrol suppresses angiotensin II type I recep- various vascular cells.54
tor (AT1R) expression in vascular smooth muscle cells (VSMCs), The role of SIRT1 in endothelial activation is less controversial
hence preventing vessel contraction and blood pressure increase.44 than in endothelial dysfunction. Studies form our lab and others
SIRT1 deacetylates and activates eNOS in vitro and improves suggest that SIRT1 exerts anti-inflammatory effects in vascular
vascular function in aortic rings ex vivo.27 Furthermore, endothe- endothelial cells (Fig. 1).46,55-57 SIRT1 deacetylates RelA/p65 at
lial overexpression of SIRT1 prevents atherosclerosis by improv- K310 and suppresses its binding to naked DNA in human aortic
ing vascular function in atherosclerosis-prone ApoE-/- mice kept endothelial cells (HAECs), therefore interfering with a crucial
on a high-fat diet.45 Conversely, organ chamber experiments step in NFκB signaling activation and reducing the expression of
using aortic rings of ApoE-/- SIRT1+/+ and ApoE-/- SIRT1+/- mice endothelial adhesion molecules.46 AcK310 deacetylation of RelA/
kept on a high-cholesterol diet, demonstrated no difference in p65 by SIRT1 has also been observed in human epithelial lung
endothelium-dependent vascular function.46 Several reasons may cells, myeloid cells and microglia.30,58,59 Importantly, suppression

www.landesbioscience.com Cell Cycle 641


Downloaded by [178.131.172.15] at 12:25 24 July 2015

©2011L
andesBiosc
ienc
e.
Figure 1. SIRT1 prevents endothelial adhesion molecule expression by suppressing the NFκB signaling pathway. Inflammatory stimuli such as TNF-α
or oxLDL lead to activation of endothelial cells, which in turn activate inflammatory signaling cascades and induce the expression of adhesion mol-

Donotdi
str
ibut
e.
ecules, such as ICAM-1, VCAM-1 and P-Selectin. These adhesion molecules attract and bind to leucocytes, thereby promoting their transmigration into
the arterial intima. SIRT1 diminishes this inflammatory activation by suppressing the NFκB signaling pathway and hence reducing the expression of
adhesion molecules.

of NFκB signaling by SIRT1 also plays an anti-atherogenic role exerts athero-protective functions in VSMCs by stabilizing
in vivo: Intra peritoneal injection of lipopolysaccharide to boost plaques.
NFκB signaling leads to increased aortic endothelial expression While many studies show that SIRT1 can fine-tune the
of ICAM-1 and VCAM-1 in ApoE-/- SIRT1+/- compared to ApoE‑/- expression RelA/p65 target genes by deacetylating RelA/p65 in
SIRT1+/+ mice.46 different cell lines, a recent study demonstrates that RelA/p65
binds to a consensus NFκB binding site at the SIRT1 promoter
SIRT1 in Vascular Smooth Muscle Cells and induces its expression in an inflammatory environment in
Stabilizes Plaques the rat aortic smooth muscle cell line A7r5.64 Enhanced SIRT1
expression under these conditions may act as a rescue mechanism
Vascular smooth muscle cells (VSCMs) compose the media of in response to inflammation.
intact arteries, but in atherogenesis VSMCs are also found in the
intima of atherosclerotic plaques. VSMC apoptosis contributes SIRT1 Decreases Cholesterol Uptake and
to plaque destabilization, but on the other hand VSMCs are the Macrophage Foam Cell Formation
only vascular cells that express stabilizing matrix components of
the fibrous cap, thereby hindering plaque rupture. Stable plaques Infiltration of monocyte-derived macrophages into the suben-
have a high VSMC content, whereas unstable plaques contain dothelial space is a key step in atherogenesis. These invading
fewer VSMCs and more macrophages.60,61 Overexpression of macrophages release and respond to inflammatory mediators
SIRT1 and nicotinamide phosphoribosyltransferase (Nampt) in and interact with cytotoxic and helper T cells. While in vitro
VSMCs increases SIRT1 activity, suppresses p21 and increases incubation of monocytes or macrophages with native LDL does
the replicative lifespan by converting them into senescence-resis- not lead to cholesterol accumulation, incubation with modified
tant cells.62 The tissue inhibitor of metalloproteinase 3 (TIMP3) LDL leads to a quick accumulation of cholesterol.65 The active
is an important inhibitor of metalloproteinases, which digest uptake of modified LDL, mainly oxLDL, is mediated by scav-
the extracellular matrix. SIRT1 silencing in VSMCs reduces enger receptors, including scavenger receptor A (SR-A), CD36,
TIMP3 expression, while SIRT1 overexpression increases Lectin-like oxLDL receptor 1 (Lox-1) and scavenger receptor for
TIMP3 promoter activity.63 These studies indicate that SIRT1 phosphatidylserine and oxidized lipoprotein/CXC chemokine

642 Cell Cycle Volume 10 Issue 4


Downloaded by [178.131.172.15] at 12:25 24 July 2015

©2011L
andesBiosc
ienc
e.
Figure 2. Macrophage SirT1 reduces inflammation, diminishes cholesterol uptake and promotes reverse cholesterol transport. Plaque macrophages
contribute to atherosclerotic disease progression by triggering an inflammatory environment, mediating macrophage foam cell formation and
necrotic core expansion. SirT1 suppresses NFκB signaling activation, thereby reducing the expression of inflammatory molecules such as TNF-α, IL-1β,

Donotdi
str
ibut
e.
IL-6 and MCP-1. SirT1 diminishes the expression of Lox-1, a receptor for oxLDL that promotes macrophage foam cell formation. Furthermore, SirT1
stimulates LXr-mediated ABCA1 expression and hence promotes reverse cholesterol transport that protects the cell from excessive cholesterol ac-
cumulation.

ligand 16 (SR-PSOX/CXCL16). Uncontrolled accumulation of SIRT1 Promotes Reverse Cholesterol Transport


modified LDL leads to the formation of lipid droplets and foam in Macrophages
cell formation.66
SIRT1 regulates both inflammatory processes and cholesterol Mammalian cells are unable to degrade the sterol ring of cho-
metabolism in macrophages (Fig. 2). SIRT1 suppresses the expres- lesterol, and most excess sterols can only be eliminated from the
sion of the scavenger receptor Lox-1 in macrophages, reduces the body by biliary excretion. Therefore, macrophages and other
uptake of oxLDL and prevents macrophage foam cell formation.67 cell types have to export cholesterol to extracellular acceptors,
Bone marrow transplantation experiments confirmed that mac- mainly HDLs, for transport to the liver. This reverse cholesterol
rophage-derived SIRT1 is crucial to prevent atherosclerosis.67 In transport involves many enzymes and receptors, such as liver X
an inflammatory context, it prevents fatty acid-induced inflam- receptors (LXRs) and their binding partners retinoid X receptor
mation,68 reduces the expression of many pro-inflammatory mol- (RXRs), peroxisome-proliferator activated receptors (PPARs),
ecules such as TNF-α, MCP-1 and interleukins,59,69 modulates the ATP-binding cassette (ABC) transporters ABCA1 and
insulin sensitivity,70 and diminishes cyclooxygenase 2 (COX-2) ABCG1 as well as the scavenger receptor SR-BI. Dysfunctional
expression and prostaglandin E2 (PGE2) secretion.71 Moreover, reverse cholesterol transport mechanisms may lead to excessive
SIRT1-deficient macrophages display NFκB hyperacetylation accumulation of cholesterol in the cell and therefore stimulate
resulting in enhanced expression of various pro-inflammatory foam cell formation and disease progression.73
genes as well as increased accumulation of activated macrophages Li et al. demonstrated that SIRT1 directly deacetylates
in the liver and adipose tissue in mice fed a high-fat diet.59 Other and thereby regulates the transcriptional activity of LXRα, an
studies showed that macrophage-derived SIRT1 regulates the important regulator of lipid homeostasis and inflammation.31
expression of matrix metalloproteinases (MMP) and the MMP- Activation of LXRα leads to expression of ABCA1 that regulates
regulator TIMP3 under pathological conditions.63,72 These find- the efflux of cholesterol into pre-βHDL particles. Indeed, Li et al.
ings suggest that SIRT1 does not only regulate the expression showed that cholesterol efflux from SIRT1+/+ is higher than from
of scavenger receptors cholesterol uptake and anti-inflammatory SIRT1-/- peritoneal macrophages ex vivo (Fig. 2). Furthermore,
events in macrophages; SIRT1 also diminishes the expression of the authors reported that the SIRT1 inhibitor NAM inhibits
MMPs, thereby promoting plaque stability. cholesterol efflux in the human monocytic cell line THP-1.31

www.landesbioscience.com Cell Cycle 643


Downloaded by [178.131.172.15] at 12:25 24 July 2015

Figure 3. Endothelial SIRT1 suppresses NFκB signaling-mediated tissue factor expression and arterial thrombosis. Release of endothelial tissue factor

©2011L
andesBiosc
ienc
e.
into the blood during plaque rupture activates the coagulation cascade, leading to platelet activation and thrombus growth. By suppressing the NFκB
signaling pathway in endothelial cells, SIRT1 diminishes tissue factor expression and activation and prevents arterial thrombosis in mice.

Donotdi
str
ibut
e.
In contrast, no change in cholesterol efflux was observed in the
mouse macrophage cell line RAW 264.7 upon treatment with
thereby activating the coagulation cascade that will activate
platelets. Activated platelets then release the contents of granules,
the SIRT1 inhibitor splitomicin.67 The different inhibitors or cell which further promote platelet recruitment, adhesion, aggrega-
types that have been used may explain the controversial results. tion and activation.82
Moreover, neither NAM nor splitomicin are solely specific for It was recently demonstrated that SIRT1 may prevent athero-
SIRT1. NAM is an end-product inhibitor of the deacetylation thrombosis by downregulating the endothelial expression of tis-
reaction mediated by the NAD + -dependent sirtuins,74 and there- sue factor: Treatment of wild-type mice with the SIRT1 inhibitor
fore an inhibitor of all NAD + -dependent sirtuins and possibly splitomicin in vivo enhanced tissue factor activity and markedly
other enzymes. Splitomicin was identified as an inhibitor of the reduced the time upon coagulation in a photochemical vascular
Saccharomyces cerevisiae Sir2p.75 Although splitomicin shows injury model (Fig. 3).83 In vitro, SIRT1 diminishes tissue factor
weak inhibitory potential towards human SIRT1,76,77 it has been expression by deacetylating RelA/p65 and suppressing NFκB sig-
used by different groups to inhibit human or mouse SIRT1.78-81 naling in HAECs. SIRT1 inhibition or siRNA-mediated SIRT1
Future studies using different specific SIRT1 inhibitors and acti- silencing in HAECs stimulated with inflammatory mediators
vators as well as genetic approaches are necessary to clarify the increased tissue factor expression.83 Patients with atherosclero-
role of SIRT1 in cholesterol efflux in vivo. sis have elevated levels of tissue factor84 and even higher con-
centrations are measured in the area around the culprit lesion
SIRT1 Inhibits Endothelial Tissue Factor Expression in patients with unstable angina or acute myocardial infarction
and Coagulation compared to patients with stable angina.85,86 The recent findings
showing that SIRT1 reduces thrombosis highlight the need for
Atherothrombosis is the primary cause of myocardial infarction evaluating pharmacological SIRT1 activation in patients with
and stroke, which are the main causes of morbidity and mortality acute coronary syndromes.
in developed countries. The main trigger of thrombosis is rupture
of a vulnerable atherosclerotic plaque. Once an atherosclerotic Perspective
plaque ruptures, highly thrombogenic material attracts platelets
to the site of injury, thereby inducing platelet aggregation and Many studies demonstrate that SIRT1 exhibits anti-inflammatory
an increase of the nascent thrombus. Tissue factor (coagulation properties both in vitro (e.g., fatty acid-induced inflammation), in
factor III) is abundantly expressed by various cells in atheroscle- vivo (e.g., myeloid deletion, atherosclerosis, sustainment of nor-
rotic plaques and released into the blood during plaque rupture, mal immune function in knock-out mice) as well as in clinical

644 Cell Cycle Volume 10 Issue 4


Downloaded by [178.131.172.15] at 12:25 24 July 2015

©2011L
andesBiosc
ienc
e.
Figure 4. Pharmacological SIRT1 activation may exert beneficial effects on atherosclerosis and thrombosis. Several metabolic and inflammatory

Donotdi
str
ibut
e.
events may enhance atherosclerosis, such as endothelial dysfunction, endothelial activation, macrophage foam cell formation, defective reverse cho-
lesterol transport (RCT) and increased tissue factor expression. Recent studies demonstrate that SIRT1 may prevent these dysfunctions to some extent.
Several of these effects are mediated via activation of eNOS in endothelial cells and LXR in macrophages as well as via suppression of NFκB signaling
activation in various plaque cells, including endothelial cells, macrophages and smooth muscle cells.

studies (e.g., patients with chronic obstructive pulmonary dis- effects and side effects of these promising activators in vascular and
ease).46,59,67,68,83,87,88 These studies imply that SIRT1 activation other chronic inflammatory diseases. Given common pathoge-
may be a promising strategy for treating chronic inflammatory netic aspects with involvement of SIRT1 in other chronic inflam-
diseases, such as atherosclerosis and atherothrombosis (Fig. 4). matory diseases such as rheumatoid arthritis, inflammatory bowel
Indeed, several SIRT1 activators have been developed by SirtrisTM disease and type 2 diabetes, it appears worthwhile to test SIRT1
Pharmaceuticals.89 While some studies claim that these activa- activators in an expanded spectrum of therapeutic indications.
tors are specific,90-92 others demonstrated that these compounds
do not act via direct SIRT1 activation.93 According to SirtrisTM Acknowledgements
Pharmaceuticals new SIRT1-activating drugs are being developed This work was supported by grants from the Swiss National
and tested in phase IIa clinical trials in patients with type 2 diabe- Science Foundation 31-114094/1, 310030_130626/1 and the
tes, inflammation and cardiovascular disease. Future studies will University Research Priority Program “Integrative Human
be necessary to better understand the SIRT1 specificity, clinical Physiology” at the University of Zurich.
5. Avalos JL, Bever KM, Wolberger C. Mechanism of sir- 9. Tsang AW, Escalante-Semerena JC. CobB, a new
References
tuin inhibition by nicotinamide: altering the NAD(+) member of the SIR2 family of eucaryotic regulatory
1. Kuo MH, Allis CD. Roles of histone acetyltransferases cosubstrate specificity of a Sir2 enzyme. Mol Cell 2005; proteins, is required to compensate for the lack of nico-
and deacetylases in gene regulation. Bioessays 1998; 17:855-68. tinate mononucleotide: 5,6-dimethylbenzimidazole
20:615-26. 6. Rine J, Herskowitz I. Four genes responsible for a phosphoribosyltransferase activity in cobT mutants
2. Michan S, Sinclair D. Sirtuins in mammals: Insights into position effect on expression from HML and HMR in during cobalamin biosynthesis in Salmonella typhimuri-
their biological function. Biochem J 2007; 404:1-13. Saccharomyces cerevisiae. Genetics 1987; 116:9-22. um LT2. J Biol Chem 1998; 273:31788-94.
3. Denu JM. The Sir 2 family of protein deacetylases. 7. Maillet L, Boscheron C, Gotta M, Marcand S, Gilson 10. Brachmann CB, Sherman JM, Devine SE, Cameron
Curr Opin Chem Biol 2005; 9:431-40. E, Gasser SM. Evidence for silencing compartments EE, Pillus L, Boeke JD. The SIR2 gene family, con-
4. Tanner KG, Landry J, Sternglanz R, Denu JM. Silent within the yeast nucleus: A role for telomere proxim- served from bacteria to humans, functions in silencing,
information regulator 2 family of NAD- dependent ity and Sir protein concentration in silencer-mediated cell cycle progression and chromosome stability. Genes
histone/protein deacetylases generates a unique prod- repression. Genes Dev 1996; 10:1796-811. Dev 1995; 9:2888-902.
uct, 1-O-acetyl-ADP-ribose. Proc Natl Acad Sci USA 8. Gottlieb S, Esposito RE. A new role for a yeast tran- 11. Frye RA. Characterization of five human cDNAs with
2000; 97:14178-82. scriptional silencer gene, SIR2, in regulation of recom- homology to the yeast SIR2 gene: Sir2-like proteins
bination in ribosomal DNA. Cell 1989; 56:771-6. (sirtuins) metabolize NAD and may have protein
ADP-ribosyltransferase activity. Biochem Biophys Res
Commun 1999; 260:273-9.

www.landesbioscience.com Cell Cycle 645


12. Blander G, Guarente L. The Sir2 family of protein 34. Thurberg BL, Collins T. The nuclear factor kappaB/ 56. Gracia-Sancho J, Villarreal G Jr, Zhang Y, Garcia-
deacetylases. Annu Rev Biochem 2004; 73:417-35. inhibitor of kappaB autoregulatory system and athero- Cardena G. Activation of SIRT1 by resveratrol induces
13. Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 sclerosis. Curr Opin Lipidol 1998; 9:387-96. KLF2 expression conferring an endothelial vasoprotec-
complex and SIR2 alone promote longevity in 35. Knowles JW, Reddick RL, Jennette JC, Shesely EG, tive phenotype. Cardiovasc Res 2010; 85:514-9.
Saccharomyces cerevisiae by two different mechanisms. Smithies O, Maeda N. Enhanced atherosclerosis and 57. Csiszar A, Labinskyy N, Podlutsky A, Kaminski PM,
Genes Dev 1999; 13:2570-80. kidney dysfunction in eNOS(-/-)Apoe(-/-) mice are Wolin MS, Zhang C, et al. Vasoprotective effects of
14. Imai S, Armstrong CM, Kaeberlein M, Guarente L. ameliorated by enalapril treatment. J Clin Invest 2000; resveratrol and SIRT1: Attenuation of cigarette smoke-
Transcriptional silencing and longevity protein Sir2 is 105:451-8. induced oxidative stress and proinflammatory phe-
an NAD-dependent histone deacetylase. Nature 2000; 36. Chien S. Effects of disturbed flow on endothelial cells. notypic alterations. Am J Physiol Heart Circ Physiol
403:795-800. Ann Biomed Eng 2008; 36:554-62. 2008; 294:2721-35.
15. Lin SJ, Defossez PA, Guarente L. Requirement of NAD 37. Chen Z, Peng IC, Cui X, Li YS, Chien S, Shyy JY. 58. Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H,
and SIR2 for life-span extension by calorie restriction in Shear stress, SIRT1 and vascular homeostasis. Proc Natl Yi S, et al. SIRT1 protects against microglia-dependent
Saccharomyces cerevisiae. Science 2000; 289:2126-8. Acad Sci USA 2010; 107:10268-73. amyloid-beta toxicity through inhibiting NFkappaB
16. Lin SJ, Ford E, Haigis M, Liszt G, Guarente L. Calorie 38. Kawashima S, Yokoyama M. Dysfunction of endotheli- signaling. J Biol Chem 2005; 280:40364-74.
restriction extends yeast life span by lowering the level al nitric oxide synthase and atherosclerosis. Arterioscler 59. Schug TT, Xu Q, Gao H, Peres-da-Silva A, Draper
of NADH. Genes Dev 2004; 18:12-6. Thromb Vasc Biol 2004; 24:998-1005. DW, Fessler MB, et al. Myeloid deletion of SIRT1
17. Lin SJ, Kaeberlein M, Andalis AA, Sturtz LA, Defossez 39. Forstermann U. Nitric oxide and oxidative stress in induces inflammatory signaling in response to environ-
PA, Culotta VC, et al. Calorie restriction extends vascular disease. Pflugers Arch 2010; 459:923-39. mental stress. Mol Cell Biol 2010.
Saccharomyces cerevisiae lifespan by increasing respira- 40. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz 60. Clarke M, Bennett M. Defining the role of vascular
tion. Nature 2002; 418:344-8. A, Freeman BA, et al. Endothelial regulation of vaso- smooth muscle cell apoptosis in atherosclerosis. Cell
18. Tissenbaum HA, Guarente L. Increased dosage of a motion in apoE-deficient mice: Implications for inter- Cycle 2006; 5:2329-31.
sir-2 gene extends lifespan in Caenorhabditis elegans. actions between peroxynitrite and tetrahydrobiopterin. 61. Weissberg PL, Clesham GJ, Bennett MR. Is vascular
Nature 2001; 410:227-30. Circulation 2001; 103:1282-8. smooth muscle cell proliferation beneficial? Lancet
19. Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, 41. Mueller CF, Laude K, McNally JS, Harrison DG. 1996; 347:305-7.
Tatar M, et al. Sirtuin activators mimic caloric restric- ATVB in focus: Redox mechanisms in blood vessels. 62. Ho C, van der Veer E, Akawi O, Pickering JG. SIRT1
tion and delay ageing in metazoans. Nature 2004; Arterioscler Thromb Vasc Biol 2005; 25:274-8. markedly extends replicative lifespan if the NAD+ salvage
Downloaded by [178.131.172.15] at 12:25 24 July 2015

430:686-9. 42. Stokes KY, Clanton EC, Russell JM, Ross CR, Granger pathway is enhanced. FEBS Lett 2009; 583:3081-5.
20. Rogina B, Helfand SL. Sir2 mediates longevity in the DN. NAD(P)H oxidase-derived superoxide mediates 63. Cardellini M, Menghini R, Martelli E, Casagrande V,
fly through a pathway related to calorie restriction. Proc hypercholesterolemia-induced leukocyte-endothelial Marino A, Rizza S, et al. TIMP3 is reduced in athero-
Natl Acad Sci USA 2004; 101:15998-6003. cell adhesion. Circ Res 2001; 88:499-505. sclerotic plaques from subjects with type 2 diabetes and
21. Colman RJ, Anderson RM, Johnson SC, Kastman EK, 43. Deanfield JE, Halcox JP, Rabelink TJ. Endothelial increased by SirT1. Diabetes 2009; 58:2396-401.
Kosmatka KJ, Beasley TM, et al. Caloric restriction function and dysfunction: Testing and clinical rel- 64. Zhang HN, Li L, Gao P, Chen HZ, Zhang R, Wei
delays disease onset and mortality in rhesus monkeys. evance. Circulation 2007; 115:1285-95. YS, et al. Involvement of the p65/RelA subunit of
Science 2009; 325:201-4. 44. Miyazaki R, Ichiki T, Hashimoto T, Inanaga K, NFkappaB in TNFalpha-induced SIRT1 expression

©2011L
andesBi
22. Kaeberlein M. Lessons on longevity from budding
yeast. Nature 2010; 464:513-9. osc
ienc
e.
23. Kaeberlein M, Powers RW, 3rd. Sir2 and calorie restric-
Imayama I, Sadoshima J, et al. SIRT1, a longevity gene,
downregulates angiotensin II type 1 receptor expression
in vascular smooth muscle cells. Arterioscler Thromb
in vascular smooth muscle cells. Biochem Biophys Res
Commun 2010; 397:569-75.
65. Goldstein JL, Ho YK, Basu SK, Brown MS. Binding
site on macrophages that mediates uptake and degrada-

Donotdi
str
ibut
e.
tion in yeast: A skeptical perspective. Ageing Res Rev Vasc Biol 2008; 28:1263-9.
2007; 6:128-40. 45. Zhang QJ, Wang Z, Chen HZ, Zhou S, Zheng W, tion of acetylated low density lipoprotein, producing
24. Baur JA, Chen D, Chini EN, Chua K, Cohen HY, Liu G, et al. Endothelium-specific overexpression of massive cholesterol deposition. Proc Natl Acad Sci USA
de Cabo R, et al. Dietary restriction: Standing up for class III deacetylase SIRT1 decreases atherosclerosis in 1979; 76:333-7.
sirtuins. Science 2010; 329:1012-3. apolipoprotein E-deficient mice. Cardiovasc Res 2008; 66. Steinberg D. Atherogenesis in perspective:
25. Fontana L, Partridge L, Longo VD. Extending healthy 80:191-9. Hypercholesterolemia and inflammation as partners in
life span—from yeast to humans. Science 2010; 46. Stein S, Schafer N, Breitenstein A, Besler C, Winnik S, crime. Nat Med 2002; 8:1211-7.
328:321-6. Lohmann C, et al. SIRT1 reduces endothelial activation 67. Stein S, Lohmann C, Schäfer N, Hofmann J, Rohrer
26. Picard F, Kurtev M, Chung N, Topark-Ngarm A, without affecting vascular function in ApoE-/- mice. L, Besler C, et al. SIRT1 decreases Lox-1-mediated
Senawong T, Machado De Oliveira R, et al. Sirt1 pro- Aging (Albany NY) 2010; 2:353-60. foam cell formation in atherogenesis. Eur Heart J 2010;
motes fat mobilization in white adipocytes by repress- 47. Pirola L, Frojdo S. Resveratrol: one molecule, many PMID: 20418343; DOI: 10.1093/eurheartj/ehq107.
ing PPAR-gamma. Nature 2004; 429:771-6. targets. IUBMB Life 2008; 60:323-32. 68. Yang Z, Kahn BB, Shi H, Xue BZ. Macrophage alpha1
27. Mattagajasingh I, Kim CS, Naqvi A, Yamamori T, 48. Beher D, Wu J, Cumine S, Kim KW, Lu SC, Atangan AMP-activated protein kinase (alpha1AMPK) antago-
Hoffman TA, Jung SB, et al. SIRT1 promotes endo- L, et al. Resveratrol is not a direct activator of SIRT1 nizes fatty acid-induced inflammation through SIRT1.
thelium-dependent vascular relaxation by activating enzyme activity. Chem Biol Drug Des 2009; 74:619-24. J Biol Chem 2010; 285:19051-9.
endothelial nitric oxide synthase. Proc Natl Acad Sci 49. Milne JC, Denu JM. The Sirtuin family: Therapeutic 69. Shen Z, Ajmo JM, Rogers CQ, Liang X, Le L, Murr
USA 2007; 104:14855-60. targets to treat diseases of aging. Curr Opin Chem Biol MM, et al. Role of SIRT1 in regulation of LPS- or
28. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane 2008; 12:11-7. two ethanol metabolites-induced TNFalpha produc-
H, Lerin C, Daussin F, et al. Resveratrol improves 50. Yang Z, Ming XF. The vascular SIRTainty. Aging tion in cultured macrophage cell lines. Am J Physiol
mitochondrial function and protects against metabolic (Albany NY) 2010; 2:331-2. Gastrointest Liver Physiol 2009; 296:1047-53.
disease by activating SIRT1 and PGC-1alpha. Cell 51. Collins T, Read MA, Neish AS, Whitley MZ, Thanos 70. Yoshizaki T, Schenk S, Imamura T, Babendure JL,
2006; 127:1109-22. D, Maniatis T. Transcriptional regulation of endothe- Sonoda N, Bae EJ, et al. SIRT1 inhibits inflamma-
29. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman lial cell adhesion molecules: NFkappaB and cytokine- tory pathways in macrophages and modulates insulin
BM, Puigserver P. Nutrient control of glucose homeo- inducible enhancers. FASEB J 1995; 9:899-909. sensitivity. Am J Physiol Endocrinol Metab 2010;
stasis through a complex of PGC-1alpha and SIRT1. 298:419-28.
52. Gareus R, Kotsaki E, Xanthoulea S, van der Made I,
Nature 2005; 434:113-8. Gijbels MJ, Kardakaris R, et al. Endothelial cell-specific 71. Zhang R, Chen HZ, Liu JJ, Jia YY, Zhang ZQ, Yang
30. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones NFkappaB inhibition protects mice from atherosclero- RF, et al. SIRT1 suppresses activator protein-1 tran-
DR, Frye RA, et al. Modulation of NFkappaB- sis. Cell Metab 2008; 8:372-83. scriptional activity and cyclooxygenase-2 expression in
dependent transcription and cell survival by the SIRT1 macrophages. J Biol Chem 2010; 285:7097-110.
53. Wood KM, Cadogan MD, Ramshaw AL, Parums DV.
deacetylase. EMBO J 2004; 23:2369-80. The distribution of adhesion molecules in human ath- 72. Nakamaru Y, Vuppusetty C, Wada H, Milne JC, Ito M,
31. Li X, Zhang S, Blander G, Tse JG, Krieger M, Guarente erosclerosis. Histopathology 1993; 22:437-44. Rossios C, et al. A protein deacetylase SIRT1 is a nega-
L. SIRT1 deacetylates and positively regulates the tive regulator of metalloproteinase-9. FASEB J 2009;
54. Zernecke A, Shagdarsuren E, Weber C. Chemokines in
nuclear receptor LXR. Mol Cell 2007; 28:91-106. 9:2810-9; PMID: 19376817.
atherosclerosis: An update. Arterioscler Thromb Vasc
32. Tontonoz P, Spiegelman BM. Fat and Beyond: The Biol 2008; 28:1897-908. 73. Li AC, Glass CK. The macrophage foam cell as a target
Diverse Biology of PPARgamma. Annu Rev Biochem for therapeutic intervention. Nat Med 2002; 8:1235-42.
55. Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P,
2008; 77:289-312. Losonczy G, et al. Anti-oxidative and anti-inflammato- 74. Bitterman KJ, Anderson RM, Cohen HY, Latorre-
33. Kim HJ, Park KG, Yoo EK, Kim YH, Kim YN, Kim ry vasoprotective effects of caloric restriction in aging: Esteves M, Sinclair DA. Inhibition of silencing and
HS, et al. Effects of PGC-1alpha on TNFalpha-induced Role of circulating factors and SIRT1. Mech Ageing accelerated aging by nicotinamide, a putative negative
MCP-1 and VCAM-1 expression and NFkappaB acti- Dev 2009; 130:518-27. regulator of yeast sir2 and human SIRT1. J Biol Chem
vation in human aortic smooth muscle and endothelial 2002; 277:45099-107.
cells. Antioxid Redox Signal 2007; 9:301-7.

646 Cell Cycle Volume 10 Issue 4


75. Bedalov A, Gatbonton T, Irvine WP, Gottschling DE, 83. Breitenstein A, Stein S, Holy EW, Camici GG, 89. Milne JC, Lambert PD, Schenk S, Carney DP, Smith
Simon JA. Identification of a small molecule inhibitor Lohmann C, Akhmedov A, et al. Sirt1 inhibition JJ, Gagne DJ, et al. Small molecule activators of SIRT1
of Sir2p. Proc Natl Acad Sci USA 2001; 98:15113-8. induces in vivo arterial thrombosis and tissue fac- as therapeutics for the treatment of type 2 diabetes.
76. Hirao M, Posakony J, Nelson M, Hruby H, Jung M, tor expression in activated human endothelial cells. Nature 2007; 450:712-6.
Simon JA, et al. Identification of selective inhibitors Cardiovasc Res 2010; 89:464-72; PMID: 20978007. 90. Yamazaki Y, Usui I, Kanatani Y, Matsuya Y, Tsuneyama
of NAD+-dependent deacetylases using phenotypic 84. Reilly MP, Rohatgi A, McMahon K, Wolfe ML, Pinto K, Fujisaka S, et al. Treatment with SRT1720, a
screens in yeast. J Biol Chem 2003; 278:52773-82. SC, Rhodes T, et al. Plasma cytokines, metabolic syn- SIRT1 Activator, Ameliorates Fatty Liver with Reduced
77. Posakony J, Hirao M, Stevens S, Simon JA, Bedalov A. drome and atherosclerosis in humans. J Investig Med Expression of Lipogenic Enzymes in MSG Mice. Am J
Inhibitors of Sir2: Evaluation of splitomicin analogues. 2007; 55:26-35. Physiol Endocrinol Metab 2009.
J Med Chem 2004; 47:2635-44. 85. Annex BH, Denning SM, Channon KM, Sketch MH 91. Funk JA, Odejinmi S, Schnellmann RG. SRT1720
78. Prozorovski T, Schulze-Topphoff U, Glumm R, Jr, Stack RS, Morrissey JH, et al. Differential expression induces mitochondrial biogenesis and rescues mito-
Baumgart J, Schroter F, Ninnemann O, et al. Sirt1 of tissue factor protein in directional atherectomy speci- chondrial function after oxidant injury in renal proximal
contributes critically to the redox-dependent fate of mens from patients with stable and unstable coronary tubule cells. J Pharmacol Exp Ther 2010; 333:593-601.
neural progenitors. Nat Cell Biol 2008; 10:385-94. syndromes. Circulation 1995; 91:619-22. 92. Dai H, Kustigian L, Carney D, Case A, Considine T,
79. Kim EJ, Kho JH, Kang MR, Um SJ. Active regulator of 86. Ardissino D, Merlini PA, Ariens R, Coppola R, Hubbard BP, et al. SIRT1 activation by small mol-
SIRT1 cooperates with SIRT1 and facilitates suppres- Bramucci E, Mannucci PM. Tissue-factor antigen and ecules—kinetic and biophysical evidence for direct
sion of p53 activity. Mol Cell 2007; 28:277-90. activity in human coronary atherosclerotic plaques. interaction of enzyme and activator. J Biol Chem 2010.
80. Biacsi R, Kumari D, Usdin K. SIRT1 inhibition allevi- Lancet 1997; 349:769-71. 93. Pacholec M, Bleasdale JE, Chrunyk B, Cunningham
ates gene silencing in Fragile X mental retardation 87. Sequeira J, Boily G, Bazinet S, Saliba S, He X, Jardine D, Flynn D, Garofalo RS, et al. SRT1720, SRT2183,
syndrome. PLoS Genet 2008; 4:1000017. K, et al. sirt1-null mice develop an autoimmune-like SRT1460 and resveratrol are not direct activators of
81. Hou X, Xu S, Maitland-Toolan KA, Sato K, Jiang B, condition. Exp Cell Res 2008; 314:3069-74. SIRT1. J Biol Chem 2010; 285:8340-51.
Ido Y, et al. SIRT1 regulates hepatocyte lipid metabo- 88. Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I.
lism through activating AMP-activated protein kinase. SIRT1, an antiinflammatory and antiaging protein, is
J Biol Chem 2008; 283:20015-26. decreased in lungs of patients with chronic obstructive
82. Mackman N. Triggers, targets and treatments for pulmonary disease. Am J Respir Crit Care Med 2008;
thrombosis. Nature 2008; 451:914-8. 177:861-70.
Downloaded by [178.131.172.15] at 12:25 24 July 2015

©2011L
andesBiosc
ienc
e.
Donotdi
str
ibut
e.

www.landesbioscience.com Cell Cycle 647

You might also like